US20190046490A1 - Methods of treating lactose intolerance - Google Patents

Methods of treating lactose intolerance Download PDF

Info

Publication number
US20190046490A1
US20190046490A1 US16/078,555 US201716078555A US2019046490A1 US 20190046490 A1 US20190046490 A1 US 20190046490A1 US 201716078555 A US201716078555 A US 201716078555A US 2019046490 A1 US2019046490 A1 US 2019046490A1
Authority
US
United States
Prior art keywords
linoleic acid
acid
conjugated linoleic
patient
lct
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US16/078,555
Other languages
English (en)
Inventor
Marie McNulty
Francesca Viti
Salvatore Bellinvia
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Nogra Pharma Ltd
Original Assignee
Nogra Pharma Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Nogra Pharma Ltd filed Critical Nogra Pharma Ltd
Priority to US16/078,555 priority Critical patent/US20190046490A1/en
Publication of US20190046490A1 publication Critical patent/US20190046490A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/20Carboxylic acids, e.g. valproic acid having a carboxyl group bound to a chain of seven or more carbon atoms, e.g. stearic, palmitic, arachidic acids
    • A61K31/201Carboxylic acids, e.g. valproic acid having a carboxyl group bound to a chain of seven or more carbon atoms, e.g. stearic, palmitic, arachidic acids having one or two double bonds, e.g. oleic, linoleic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system

Definitions

  • lactose deficiency Approximately 70% of the world's population has primary lactase deficiency. The percentage of lactose deficiency varies according to ethnicity and is related to the use of dairy products in the diet reaching up to 20% of North European, 40% of Mediterranean European, 80% of Africans, and 90% of Asian population. No curative treatments for primary lactose intolerance are currently available, with typical treatments for lactose intolerance including lactose exclusion (leading to nutritional impairment) or expansive regimen such as the use of lactose deficient milk or lactase supplementation. In the United States alone, the annual financial burden of lactose intolerance is estimated to be nearly 2 billion dollars.
  • PPARs Peroxisome Proliferator Activated Receptors
  • PPARs are members of the nuclear hormone receptor super family, which are ligand-activated transcription factors regulating gene expression. PPARs play a role in the regulation of cell differentiation, development and metabolism of higher organisms.
  • Described herein are methods for treating and/or ameliorating lactose intolerance or lactase deficiency in a patient in need thereof, the method comprising administering a composition comprising an isolated fatty acid to the patient. Also described herein are methods for stimulating lactase gene expression in a patient in need thereof, comprising administering a composition comprising an isolated fatty acid to said patient, and methods for treating diarrhea, abdominal pain and/or bloating after lactose ingestion in a lactose intolerant patient in need thereof, comprising administering a composition comprising an isolated fatty acid.
  • the disclosure is directed to a method for treating and/or ameliorating lactose intolerance or lactase deficiency in a patient in need thereof, where the method includes administering to the patient a composition consisting essentially of a fatty acid, for example, a conjugated linoleic acid.
  • a fatty acid is a naturally occurring fatty acid, for example, a naturally occurring conjugated linoleic acid.
  • the administering may be before, after, or substantially concurrent with the consumption of a food that includes a dairy product.
  • the methods include administering a composition that includes a fatty acid daily, weekly, or as needed over a time period of 3 months, 6 months, 1 year, or more.
  • a patient e.g., a human patient
  • the fatty acid is a conjugated linoleic acid, e.g., trans-10, cis-12 conjugated linoleic acid isomer, cis-9, trans-11 conjugated linoleic acid isomer, or a mixture thereof.
  • a food product that includes a therapeutically effective amount of a fatty acid to ameliorate lactose intolerance in a patient.
  • the food product includes a therapeutically effective amount of a fatty acid to ameliorate lactose intolerance in a patient and, optionally, a dairy component, e.g., whey, milk, cheese or cream.
  • the fatty acid is a conjugated linoleic acid, e.g. the trans-10, cis-12 conjugated linoleic acid isomer, the cis-9, trans-11 conjugated linoleic acid isomer, or a mixture thereof.
  • a food product comprising a fatty acid, for example, a conjugated linoleic acid, in an amount significantly greater than a naturally occurring amount of a fatty acid, for example, a naturally occurring amount of a conjugated linoleic acid, in the food product, e.g., wherein the amount of the fatty acid (for example, a conjugated linoleic acid) is about 5%, about 10%, about 50%, about 100%, or more than about 100% by weight greater than a naturally occurring amount of the fatty acid (for example, a naturally occurring amount of a conjugated linoleic acid) in the food product.
  • a fatty acid for example, a conjugated linoleic acid
  • the food product includes a conjugated linoleic acid where the conjugated linoleic acid is a trans-10, cis-12 conjugated linoleic acid isomer, a cis-9, trans-11 conjugated linoleic acid isomer, or a mixture thereof.
  • the disclosure is directed to nutraceutical compositions that include a therapeutically effective amount of a fatty acid, for example, a conjugated linoleic acid, where the therapeutically effective amount of the fatty acid, for example, the therapeutically effective amount of a conjugated linoleic acid, substantially prevents, ameliorates, or treats lactose intolerance in a human patient when orally administered or consumed by the patient.
  • a therapeutically effective amount of a fatty acid for example, a conjugated linoleic acid
  • a nutraceutical composition includes a conjugated linoleic acid, where the conjugated linoleic acid is a trans-10, cis-12 conjugated linoleic acid isomer, a cis-9, trans-11 conjugated linoleic acid isomer, or a mixture thereof.
  • a pharmaceutical formulation of the disclosure includes a fatty acid, a pharmaceutically acceptable filler, and an enteric coating.
  • a pharmaceutical formulation includes a fatty acid that is a conjugated linoleic acid.
  • a pharmaceutical formulation includes a fatty acid where the fatty acid is a trans-10, cis-12 conjugated linoleic acid isomer, a cis-9, trans-11 conjugated linoleic acid isomer, or a mixture thereof.
  • a pharmaceutical formulation of the disclosure includes a disintegrant.
  • a pharmaceutical formulation of the disclosure includes a lubricant.
  • a pharmaceutical formulation of the disclosure includes an enteric coating, where the enteric coating is about 1% to about 10%, about 5% to about 10%, about 8% to about 10%, about 8% to about 12%, about 8% to about 15%, about 8% to about 20%, about 10% to about 12%, about 10% to about 18%, or about 15% to about 20% by weight of the pharmaceutical formulation.
  • the enteric coating is ethylacrylate methacrylic acid.
  • a pharmaceutical formulation of the disclosure when orally administered to a patient, results in delivering the fatty acid to the duodenum of the patient and/or the jejunum of the patient. In some embodiments, a pharmaceutical formulation of the disclosure, when orally administered to a patient, results in release of fatty acid at a pH value of about 4.5, about 5, about 5.5, about 6, about 6.5, or about 7. In some embodiments, a pharmaceutical formulation of the disclosure, when administered to a patient, results in release of fatty acid in the gastrointestinal tract in an environment of about pH 4.5, about pH 5, about pH 5.5, about pH 6, about pH 6.5, or about pH 7.
  • a pharmaceutical formulation of the disclosure when orally administered to a patient results in amelioration or treatment of lactose intolerance or lactase deficiency in the patient.
  • a pharmaceutical formulation of the disclosure results in amelioration or treatment of lactose intolerance or lactase deficiency in the patient after the formulation is administered a defined number of times over a defined period of time, for example, after 1 time, after 2 times, after 3 times, after 4 times, after 5 times, after 6 times, after 7 times, after 8 times, after 9 times, after 10 times, or after more than 10 times over the course of 1 hour, 1 day, 1 week, or 1 month.
  • the disclosure is directed to a fatty acid, for example, linoleic acid, for example, conjugated linoleic acid, for use as a medicament, for example, for treating, preventing, managing, and/or ameliorating lactose intolerance or lactase deficiency in a patient in need thereof.
  • the disclosure is directed to a fatty acid for use in treating, preventing, managing, and/or ameliorating lactose intolerance or lactase deficiency in a patient in need thereof.
  • the fatty acid for use in treating, preventing, managing, and/or ameliorating lactose intolerance or lactase deficiency in a patient in need thereof is for use in any of the methods disclosed herein.
  • Use of a fatty acid, for example, linoleic acid, for example, conjugated linoleic acid, in the manufacture of a medicament for the treatment, prevention, management, and/or amelioration of lactose intolerance or lactase deficiency by a method described herein is also provided herein.
  • FIG. 1A depicts quantitative PCR (qPCR) analysis showing induction of LCT mRNA expression by 1 mM 3-(4′-aminophenyl)2-methoxypropionic acid (GED) in Caco-2 cells relative to unstimulated (CTL) cells (CTL v. 1 mM GED, p ⁇ 0.0001).
  • FIG. 1B depicts qPCR analysis showing induction of LCT mRNA expression by 1 ⁇ M pioglitazone (Pio) in Caco-2 cells relative to CTL cells (CTL v. 1 ⁇ M Pio, p ⁇ 0.0001). Results in FIGS. 1A and 1B represent the mean ⁇ standard error of the mean (SEM) of 4 independent experiments.
  • SEM standard error of the mean
  • the expression level measured in control cells was used as a reference in each of FIGS. 1A-1D .
  • FIG. 2A depicts the dose-effect of GED on LCT mRNA expression in Caco-2 cells, where cells were stimulated with 0.1 mM, 1 mM, or 30 mM GED, and LCT mRNA expression relative to controls (CTRL; DMEM) was determined by qRT-PCR.
  • FIG. 2B depicts the dose-effect of Pio on LCT mRNA expression in Caco-2 cells, where cells were stimulated with 0.1 ⁇ M, 1 ⁇ M, or 10 ⁇ M Pio, and LCT mRNA expression relative to controls (CTRL; DMSO) was determined by qRT-PCR.
  • Results in FIGS. 2A and 2B represent the mean ⁇ SEM of 2 to 3 independent experiments performed in triplicate (*, P ⁇ 0.05; *** P ⁇ 0.001; NS, not significant). The expression level measured in CTRL cells was used as a reference.
  • FIG. 3 is a bar graph display of LCT protein expression assessed by immunoprecipitation assay.
  • LCT protein was immunoprecipitated from Caco-2 cells either stimulated with 1 mM GED (GED) or left unstimulated (CTRL). Bars represent LCT protein signal intensity relative to ⁇ -actin signal intensity. CTRL signal was arbitrarily defined as 100%.
  • FIG. 4A is a bar graph depicting LCT activity in Caco-2 cells after stimulation with 1mM GED (GED) or no stimulation (CTRL). Results represent the mean ⁇ SEM (3 independent experiments performed in triplicate) of the percentage of LCT activity compared to the activity in CTRL cells, arbitrarily defined as 100%.
  • FIG. 4B is a bar graph depicting LCT activity in Caco-2 cells after stimulation with 1 ⁇ M Pio (Pio) or no stimulation (CTRL). Results represent the mean ⁇ SEM (3 independent experiments in triplicate) of the percentage of LCT activity compared to the activity in CTRL cells, arbitrarily defined as 100%.
  • FIG. 4C is a bar graph depicting LCT activity in Caco-2 cells after stimulation with 1 mM GED (GED1mM), 30 mM GED (GED30 mM), 30 mM 5-ASA (5ASA30 mM), or no stimulation (CTRL). Lactase activity was significantly upregulated compared to CTRL samples following stimulation with 1 mM GED (CTRL v. GED 1 mM, p ⁇ 0.005) and 30 mM GED (CTRL v. GED 30 mM, p ⁇ 0.005).
  • FIG. 5 depicts the glucose uptake capacity of Caco-2 cells after 1 mM GED (GED 1 mM) and 1 ⁇ M pioglitazone (Pio 1 ⁇ M) stimulation or no stimulation (CTRL). The result is expressed in the amount of phosphorylation of the glucose analog 2-deoxyglucose (2-DG6P) measured in the cells (pmol). NS, not significant.
  • FIG. 6A depicts the relative expression level of sucrase-isomaltase (SIM) and maltase-glucoamylase (MGAM) mRNA compared to LCT mRNA in Caco-2 cells following stimulation with a PPAR ⁇ agonist as determined by qPCR.
  • SIM sucrase-isomaltase
  • MGAM maltase-glucoamylase
  • FIG. 6B depicts the relative expression level of SIM mRNA in Caco-2 cells as determined by qPCR following stimulation with 1 mM GED (left) or 1 ⁇ M Pio (right) or left unstimulated (CTRL and DMSO). Results represent the mean ⁇ SEM (2 independent experiments performed in sextuplicate) of the fold change of expression of SIM mRNA normalized to GAPDH level. The expression level measured in control cells (arbitrarily defined as one) was used as reference. ** P ⁇ 0.01; ***P ⁇ 0.001; NS, not significant.
  • FIG. 6C depicts the relative expression level of MGAM mRNA in Caco-2 cells as determined by qPCR following stimulation with 1 mM GED (left) or 1 ⁇ M Pio (right) or left unstimulated (CTRL and DMSO). Results represent the mean ⁇ SEM (2 independent experiments performed in sextuplicate) of the fold change of expression of MGAM mRNA normalized to GAPDH level. The expression level measured in control cells (arbitrarily defined as one) was used as reference. ** P ⁇ 0.01; NS, not significant.
  • FIG. 8 is a schematic of the PPAR response element (PPRE) identified by in silico analysis in the promoter region of the human LCT gene (up to 3,000 bp upstream of the putative transcription start site) and the direct repeat 1 (DR1) and direct repeat 2 (DR2) response elements located in the region.
  • PPRE PPAR response element
  • DR1 and DR2 response elements located in the region.
  • 8 a and 8 b denote the primer pair used to amplify the genomic region encompassing the DR2 located between nucleotides ⁇ 223 to ⁇ 210.
  • FIG. 9 depicts the nucleotide sequence of the PPRE (DR1 and DR2) in the human LCT promoter gene (up to 3,000 bp upstream to the transcription start point).
  • the putative DR1's and DR2's identified in the 3,000 bp sequence of the LCT gene promoter are underlined.
  • the underlined nucleotide sequence “TAAATA” denotes a potential TATA box.
  • FIG. 9 discloses SEQ ID NO: 3.
  • FIG. 10 depicts a bar graph showing qPCR amplification signal of the 8 a - 8 b fragment in a ChIP assay from Caco-2 cells either treated with GED (GED) or not treated (CTRL). Results are expressed as fold enrichment relative to CTRL cells.
  • FIG. 11 depicts results of a luciferase gene reporter assay in Caco-2 cells transfected with a reporter construct containing the DR2 response element upstream of a luciferase gene sequence (pGL4Luc PromLCT construct) or a control construct containing a luciferase gene sequence but no upstream DR2 sequence (pGL4Luc).
  • Results represent the mean ⁇ SEM of luciferase activity normalized for protein content (2 independent experiments in triplicate) following stimulation with GED or no stimulation (CTL).
  • FIG. 12 depicts LCT mRNA expression as measured by qPCR (left) and LCT activity (right) in stably transfected PPAR ⁇ knock-down Caco-2 cells (ShPPAR) compared to stably transfected control cells (ShLuc). Results represent the mean ⁇ SEM of 3 independent experiments performed in triplicate or sextuplicate (**, P ⁇ 0.01; ***, P ⁇ 0.001).
  • FIG. 13 is a bar graph depicting the effect of the PPAR ⁇ antagonist GW9662 on GED-dependent induction of LCT mRNA expression in Caco-2 cells.
  • LCT mRNA expression was determined by qPCR. Cells were treated with GW9662 (+GW9662) or left untreated ( ⁇ GW9662) and then treated with GED (GED) or left untreated (Control). Results represent the mean ⁇ SEM (of 2 independent experiments performed in triplicate and sextuplicate) of the fold change in LCT mRNA expression, relative to cells that were not treated with GW9662 or GED (**, P ⁇ 0.01; ***, P ⁇ 0.001).
  • FIG. 15B is a graph depicting a correlation of LCT mRNA and PPAR ⁇ mRNA levels in the jejunum of weaned (squares) and not weaned (circles) rats.
  • FIG. 19A depicts the chemical structures of a series of naturally occurring PPAR ⁇ ligands.
  • FIG. 20A is a bar graph depicting LCT mRNA expression as measured by qPCR in stably transfected PPAR ⁇ knock-down Caco-2 cells (ShPPAR) and stably transfected control cells (ShLuc) not stimulated (CTRL) or stimulated with CLA at various concentrations (250 ⁇ M, 500 ⁇ M, 1000 ⁇ M).
  • FIG. 20B is a bar graph depicting LCT mRNA activity in stably transfected PPAR ⁇ knock-down Caco-2 cells (ShPPAR) and stably transfected control cells (ShLuc) not stimulated (CTRL) or stimulated with 1 mM GED (GED 1 mM) or CLA at various concentrations (250 ⁇ M, 500 ⁇ M, 1000 ⁇ M).
  • FIG. 21B is a graph depicting individual data points and mean values (horizontal bars) for LCT mRNA expression levels (normalized to GAPDH mRNA expression) in duodenal tissue of rats fed a control diet supplemented with 0.5% carboxymethyl cellulose (CMC), 30 mg/kg/day GED (GED), or 200 mg/kg/day CLA (CLA), relative to CMC controls.
  • CMC carboxymethyl cellulose
  • GED 30 mg/kg/day GED
  • CLA 200 mg/kg/day CLA
  • FIG. 21D is a graph depicting individual data points and mean values (horizontal bars) for PPAR ⁇ mRNA expression levels (normalized to GAPDH mRNA expression) in duodenal tissue of rats fed a control diet supplemented with 0.5% carboxymethyl cellulose (CMC) or 200 mg/kg/day CLA (CLA), relative to CMC controls.
  • CMC carboxymethyl cellulose
  • CLA 200 mg/kg/day CLA
  • FIG. 22A is a graph depicting the correlation between PPAR ⁇ (PPARg) and LCT (LCT) mRNA expression levels in the duodenal tissue of rats fed a control diet supplemented with 0.5% carboxymethyl cellulose.
  • FIG. 22E is a graph depicting individual data points and mean values (horizontal bars) of fold change in LCT activity in duodenal tissue of rats fed a control diet supplemented with 0.5% carboxymethyl cellulose (CMC), 30 mg/kg/day GED (GED), or 200 mg/kg/day CLA (CLA) for 5 days.
  • CMC carboxymethyl cellulose
  • GED 30 mg/kg/day GED
  • CLA 200 mg/kg/day CLA
  • FIG. 22F is a graph depicting individual data points and mean values (horizontal bars) of fold change in LCT activity in jejunal tissue of rats fed a control diet supplemented with 0.5% carboxymethyl cellulose (CMC), 30 mg/kg/day GED (GED), or 200 mg/kg/day CLA (CLA) for 5 days.
  • CMC carboxymethyl cellulose
  • GED 30 mg/kg/day GED
  • CLA 200 mg/kg/day CLA
  • compositions may also contain other active compounds providing supplemental, additional, or enhanced therapeutic functions.
  • “Individual,” “patient,” or “subject” are used interchangeably and include to any animal, including mammals, preferably mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, or primates, and most preferably humans.
  • the compounds of the invention can be administered to a mammal, such as a human, but can also be other mammals such as an animal in need of veterinary treatment, e.g., domestic animals (e.g., dogs, cats, and the like), farm animals (e.g., cows, sheep, pigs, horses, and the like) and laboratory animals (e.g., rats, mice, guinea pigs, and the like).
  • the mammal treated in the methods of the invention is desirably a mammal in whom modulation of PPAR receptors is desired.
  • “Modulation” includes antagonism (e.g., inhibition), agonism, partial antagonism and/or partial agonism.
  • terapéuticaally effective amount means the amount of the subject compound that will elicit the biological or medical response of a tissue, system, animal or human that is being sought by the researcher, veterinarian, medical doctor or other clinician.
  • the compounds of the invention are administered in therapeutically effective amounts to treat a disease.
  • a therapeutically effective amount of a compound is the quantity required to achieve a desired therapeutic and/or prophylactic effect, such as an amount which results in the prevention of or a decrease in the symptoms associated with a disease associated with PPAR receptors.
  • pharmaceutically acceptable salt(s) refers to salts of acidic or basic groups that may be present in compounds used in the present compositions.
  • Compounds included in the present compositions that are basic in nature are capable of forming a wide variety of salts with various inorganic and organic acids.
  • the acids that may be used to prepare pharmaceutically acceptable acid addition salts of such basic compounds are those that form non-toxic acid addition salts, i.e., salts containing pharmacologically acceptable anions, including but not limited to malate, oxalate, chloride, bromide, iodide, nitrate, sulfate, bisulfate, phosphate, acid phosphate, isonicotinate, acetate, lactate, salicylate, citrate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, gluconate, glucuronate, saccharate, formate, benzoate, glutamate, methanesulfonate, ethanesulfonate, benzenesulfonate, p-toluenesulfonate and pamoate (i.e., 1,1′-methylene-bis-
  • the compounds of the disclosure may contain one or more chiral centers and/or double bonds and, therefore, exist as stereoisomers, such as geometric isomers, enantiomers or diastereomers.
  • stereoisomers when used herein consist of all geometric isomers, enantiomers or diastereomers. These compounds may be designated by the symbols “R” or “S,” depending on the configuration of substituents around the stereogenic carbon atom.
  • Stereoisomers include enantiomers and diastereomers. Mixtures of enantiomers or diastereomers may be designated “( ⁇ )” in nomenclature, but the skilled artisan will recognize that a structure may denote a chiral center implicitly.
  • Individual stereoisomers of compounds of the present invention can be prepared synthetically from commercially available starting materials that contain asymmetric or stereogenic centers, or by preparation of racemic mixtures followed by resolution methods well known to those of ordinary skill in the art. These methods of resolution are exemplified by (1) attachment of a mixture of enantiomers to a chiral auxiliary, separation of the resulting mixture of diastereomers by recrystallization or chromatography and liberation of the optically pure product from the auxiliary, (2) salt formation employing an optically active resolving agent, or (3) direct separation of the mixture of optical enantiomers on chiral chromatographic columns.
  • Stereoisomeric mixtures can also be resolved into their component stereoisomers by well known methods, such as chiral-phase gas chromatography, chiral-phase high performance liquid chromatography, crystallizing the compound as a chiral salt complex, or crystallizing the compound in a chiral solvent.
  • Stereoisomers can also be obtained from stereomerically-pure intermediates, reagents, and catalysts by well known asymmetric synthetic methods.
  • Substituents around a carbon-carbon double bond alternatively can be referred to as “cis” or “trans,” where “cis” represents substituents on the same side of the double bond and “trans” represents substituents on opposite sides of the double bond.
  • the arrangement of substituents around a carbocyclic ring are designated as “cis” or “trans.”
  • the term “cis” represents substituents on the same side of the plane of the ring and the term “trans” represents substituents on opposite sides of the plane of the ring.
  • Mixtures of compounds wherein the substituents are disposed on both the same and opposite sides of plane of the ring are designated “cis/trans.”
  • the invention also embraces isotopically labeled compounds of the invention which are identical to those recited herein, except that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature.
  • isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, fluorine and chlorine, such as 2 H, 3 H, 13 C, 14 C, 15 N, 18 O, 17 O, 31 P, 32 P, 35 S, 18 F, and 36 Cl, respectively.
  • Certain isotopically-labeled disclosed compounds are useful in compound and/or substrate tissue distribution assays. Tritiated (i.e., 3 H) and carbon-14 (i.e., 14 C) isotopes are particularly preferred for their ease of preparation and detectability. Further, substitution with heavier isotopes such as deuterium (i.e., 2 H) may afford certain therapeutic advantages resulting from greater metabolic stability (e.g., increased in vivo half-life or reduced dosage requirements) and hence may be preferred in some circumstances.
  • Isotopically labeled compounds of the invention can generally be prepared by following procedures analogous to those disclosed in the e.g., Examples herein by substituting an isotopically labeled reagent for a non-isotopically labeled reagent.
  • Fatty acids include, but are not limited to, saturated fatty acids, unsaturated fatty acids, short-chain fatty acids (e.g., fatty acids with aliphatic tails of fewer than six carbons), medium-chain fatty acids (e.g., fatty acids with aliphatic tails of 6-12 carbons), long-chain fatty acids (e.g., fatty acids with aliphatic tails of 13 to 21 carbons), linoleic acid, very long chain fatty acids (e.g., fatty acids with aliphatic tails longer than 22 carbons), omega-3 fatty acids, and essential fatty acids.
  • Fatty acids also include isomers of fatty acids, for example, isomers of conjugated linoleic acid.
  • Fatty acids also include isomers of fatty acids, for example, trans and cis isomers of fatty acids.
  • Unsaturated fatty acids include, for example, but are not limited to, myristoleic acid, palmitoleic acid, sapienic acid, oleic acid, elaidic acid, vaccenic acid, linoleic acid, linoelaidic acid, ⁇ -linolenic acid, arachidonic acid, eicosapentaenoic acid, erucic acid, stearidonic acid, ⁇ -Linolenic acid, dihomo- ⁇ -linolenic acid, docosatetraenoic acid, paullinic acid, gondoic acid, gadoleic acid, eicosenoic acid, nervonic acid, mead acid, crotonic acid, eicosadienoic acid, docosadienoic acid, pinolenic acid, elostearic acid, ⁇ -eleostearic acid, eicosatrienoic acid, eicosatetrano
  • Saturated fatty acids include, for example, but are not limited to, propionic acid, butyric acid, valeric acid, caproic acid, caprylic acid, capric acid, lauric acid, myristic acid, palmitic acid, stearic acid, arachidic acid, behenic acid, lignoceric acid, cerotic acid, enanthic acid, pelargonic acid, undecylic acid, lauric acid, tridecylic acid, myristic acid, pentadecylic acid, margaric acid, nonadecylic acid, heneicosylic acid, tricosylic acid, pentacosylic acid, heptacosylic acid, montanic acid, nonacosylic acid, melissic acid, henatriacontylic acid, lacceroic acid, psyllic acid, geddic acid, ceroplastic acid, hexatriacontylic acid, heptatriacontanoic acid, and
  • Fatty acids also include stereoisomers of fatty acids and racemic mixtures of fatty acid stereoisomers, for example, stereoisomers of linoleic acid, for example, 9(5)-hydroxy-10(E),12(Z)-octadecadienoic acid (9(S)-HODE) and 9(R)-hydroxy-10(E),12(Z)-octadecadienoic acid (9(R)-HODE), and racemic mixtures of linoleic acid stereoisomers, for example, 9-hydroxyoctadecadienoic acid (9-HODE).
  • stereoisomers of linoleic acid for example, 9(5)-hydroxy-10(E),12(Z)-octadecadienoic acid (9(S)-HODE) and 9(R)-hydroxy-10(E),12(Z)-octadecadienoic acid (9(R)-HODE
  • racemic mixtures of linoleic acid stereoisomers for example, 9-
  • stereoisomers of fatty acids include, but are not limited to 13-hydroxyoctadecadienoic acid (also known as 13-HODE, 13(S)-hydroxy-9Z,11E-octadecadienoic acid, or 13(S)-HODE) and 13(R)-hydroxy-9Z,11E-octadecadienoic acid (13(R)-HODE).
  • 13-hydroxyoctadecadienoic acid also known as 13-HODE, 13(S)-hydroxy-9Z,11E-octadecadienoic acid, or 13(S)-HODE
  • 13(R)-hydroxy-9Z,11E-octadecadienoic acid 13(R)-HODE
  • a fatty acid can be, e.g., a conjugated linoleic acid.
  • Conjugated linoleic acid refers to a group of positional and geometric isomers of linoleic acid that are characterized by the presence of conjugated dienes.
  • a fatty acid can include any isomer of conjugated linoleic acid, including, e.g., the cis-9,trans-11 (c9,t11) isomer, trans-10,cis-12 (t10, c12) isomer, and trans-10,cis-11 (t10, c11) isomer.
  • Exemplary conjugated linoleic acids are represented below the structure of linoleic acid:
  • compositions comprising compounds as disclosed herein (e.g., an isolated CLA, as described above) formulated together with one or more pharmaceutically or cosmetically acceptable carriers.
  • exemplary compositions provided herein include compositions comprising essentially a CLA, as described above, and one or more pharmaceutically acceptable carriers.
  • Formulations include those suitable for oral, rectal, topical, buccal, and parenteral (e.g., subcutaneous, intramuscular, intradermal, or intravenous) administration, or for topical use, e.g., as a cosmetic product. The most suitable form of administration in any given case will depend on the degree and severity of the condition being treated and on the nature of the particular compound being used.
  • the disclosure is also directed to compositions for treating, preventing, monitoring, and/or ameliorating lactose intolerance and/or lactase deficiency that include one or more derivatives of fatty acids or products of fatty acid metabolism. In some embodiments, the disclosure is also directed to methods for treating, preventing, monitoring, and/or ameliorating lactose intolerance and/or lactase deficiency that include administering to a patient a composition that includes one or more derivatives of fatty acids or products of fatty acid metabolism.
  • Derivatives of fatty acids and products of fatty acid metabolism include, for example, hormones such as prostaglandins (for example, 15-deoxy-delta-12,14-prostaglandin J2 (15d-PGJ2)), triglycerides, phospholipids, diacyl glycerols, second messengers (for example, inositol trisphosphate), and ketone bodies.
  • hormones such as prostaglandins (for example, 15-deoxy-delta-12,14-prostaglandin J2 (15d-PGJ2)), triglycerides, phospholipids, diacyl glycerols, second messengers (for example, inositol trisphosphate), and ketone bodies.
  • Derivatives of fatty acids include derivatives of linoleic acid, for example, DCP-LA (8-[2-(2-pentyl-cyclopropylmethyl)-cyclopropyl]-octanoic acid), FR236924, and oxidized derivatives of linoleic acid, including, but not limited to, 12,13-epoxy-9-keto-(10-trans)-octadecenoic acid (EKODE).
  • DCP-LA 2-[2-(2-pentyl-cyclopropylmethyl)-cyclopropyl]-octanoic acid
  • FR236924 oxidized derivatives of linoleic acid, including, but not limited to, 12,13-epoxy-9-keto-(10-trans)-octadecenoic acid (EKODE).
  • Derivatives of fatty acids also include derivatives of arachidonic acid, including, but not limited to, 5-hydroxyicosatetraenoic acid (5-HETE), 12-hydroxyeicosatetraenoic acid (12-HETE), 15-hydroxyeicosatetraenoic acid (15-HETE), 16(R)-hydroxyeicosatetraenoic acid (16(R)-HETE), 16(S)-hydroxyeicosatetraenoic acid (16(S)-HETE), and 5(S),6(R)-Lipoxin A4, 5(S),6(R), and 15(R)-Lipoxin A4.
  • Derivatives of fatty acids also include polyethylene glycol (PEG)ylated derivatives of fatty acids, for example pegylated derivatives of linoleic acid, for example, pegylated conjugated linoleic acid.
  • PEG polyethylene glycol
  • the disclosure is directed to compositions for treating, preventing, monitoring, and/or ameliorating lactose intolerance and/or lactase deficiency that include one or more intermediate products of fatty acid metabolism, for example, intermediate products of linoleic acid metabolism.
  • the disclosure is also directed to methods for treating, preventing, monitoring, and/or ameliorating lactose intolerance and/or lactase deficiency that include administering to a patient a composition that includes one or more intermediate products of fatty acid metabolism, for example, intermediate products of linoleic acid metabolism.
  • Intermediate products of linoleic metabolism include, for example, ⁇ -linolenic acid, dihomo- ⁇ -linolenic acid, arachidonic acid, and docosatetranoic acid.
  • the disclosure is directed to compositions for treating, preventing, monitoring, and/or ameliorating lactose intolerance and/or lactase deficiency that include one or more fatty acid prodrugs, for example, a prodrug of conjugated linoleic acid.
  • the disclosure is also directed to methods for treating, preventing, monitoring, and/or ameliorating lactose intolerance and/or lactase deficiency that include administering to a patient a composition that includes one or more fatty acid prodrugs, for example, a prodrug of conjugated linoleic acid.
  • prodrug refers to a compound that is metabolized (e.g., metabolized after administration to a patient) into a pharmacologically active compound, for example, a pharmacologically active fatty acid.
  • prodrugs of conjugated linoleic acid include compounds that are metabolized to conjugated linoleic acid.
  • the disclosure is directed at least in part to treating or ameliorating lactose intolerance or a lactase deficiency (or, e.g., controlling symptoms of lactose intolerance) by administering a fatty acid, e.g., a linoleic acid, e.g., a conjugated linoleic acid to a patient (e.g., a human patient) in need thereof.
  • a fatty acid e.g., a linoleic acid, e.g., a conjugated linoleic acid
  • the disclosure is directed to methods of treating or ameliorating lactose intolerance or lactase deficiency in a patient by administering a fatty acid, e.g., a conjugated linoleic acid isomer, before, substantially simultaneously with, or after the patient ingests lactose, for example, a composition that includes lactose, for example, a food product that includes lactose.
  • a fatty acid e.g., a conjugated linoleic acid isomer
  • compositions for reducing lactose intolerance or lactase deficiency may form part of, or is used for making, a low lactose content milk or milk product, comprising a fatty acid, for example, a conjugated linoleic acid.
  • a fatty acid for example, a conjugated linoleic acid.
  • Such compositions may be or may be part of, for example, a whey product, a milk product, or a cheese product.
  • Compounds of the invention may be administered to subjects (e.g., animals and/or humans) in need of such treatment or amelioration in dosages that will provide optimal pharmaceutical efficacy.
  • subjects e.g., animals and/or humans
  • dose required for use in any particular application will vary from patient to patient, not only with respect to the particular compound or composition selected, but also with respect to the route of administration, the nature of the condition being treated, the age and condition of the patient, concurrent medication or special diets then being followed by the patient, and other factors which those skilled in the art will recognize, with the appropriate dosage ultimately being at the discretion of the attendant physician, caretaker, or patient.
  • compounds of this invention may be administered, for example, orally, topically, parenterally, by inhalation spray, or rectally in dosage unit formulations containing conventional, non-toxic, pharmaceutically acceptable carriers, adjuvants, and vehicles.
  • parenteral as used herein includes subcutaneous injections, intravenous, intramuscular, intrasternal injection, or infusion techniques.
  • a therapeutically effective amount of active component will be in the range of from about 0.1 mg/kg to about 100 mg/kg, from about 0.1 mg/kg to about 1 mg/kg, from about 0.1 mg/kg to about 10 mg/kg, from about 1 mg/kg to about 100 mg/kg, from about 1 mg/kg to 10 mg/kg, from about 10 mg/kg to about 20 mg/kg, from about 20 mg/kg to about 30 mg/kg, from about 30 mg/kg to about 40 mg/kg, from about 40 mg/kg to about 50 mg/kg, from about 50 mg/kg to about 60 mg/kg, from about 60 mg/kg to about 70 mg/kg, from about 70 mg/kg to about 80 mg/kg, from about 80 mg/kg to about 90 mg/kg, or from about 90 mg/kg to about 100 mg/kg.
  • the amount administered will depend on variables such as the type and extent of disease or indication to be treated, the overall health status of the particular patient, the relative biological efficacy of the compounds, formulations of compounds, the presence and types of excipients in the formulation, and the route of administration.
  • the initial dosage administered may be increased beyond the upper level in order to rapidly achieve the desired blood-level or tissue level, or the initial dosage may be smaller than the optimum and the daily dosage may be progressively increased during the course of treatment depending on the particular situation.
  • Human dosage can be optimized, e.g., in a conventional Phase I dose escalation study designed to run from 0.5 mg/kg to 20 mg/kg.
  • Dosing frequency can vary, depending on factors such as route of administration, dosage amount, and the disease condition being treated. Exemplary dosing frequencies are once per day, once per week and once every two weeks.
  • Formulations or compositions of the disclosure can comprise a disclosed compound and typically can also include a pharmaceutically acceptable carrier or excipient.
  • compositions of the disclosure may be administered by various means, depending on their intended use, as is well known in the art.
  • compositions of the present invention may be formulated as tablets, capsules, granules, powders or syrups.
  • formulations of the present invention may be administered parenterally as injections (intravenous, intramuscular, or subcutaneous), drop infusion preparations, enemas, or suppositories.
  • compositions of the present invention may be formulated as eyedrops or eye ointments.
  • compositions may be prepared by conventional means, and, if desired, the compositions may be mixed with any conventional additive, such as an excipient, a binder, a disintegrating agent, a lubricant, a corrigent, a solubilizing agent, a suspension aid, an emulsifying agent or a coating agent.
  • any conventional additive such as an excipient, a binder, a disintegrating agent, a lubricant, a corrigent, a solubilizing agent, a suspension aid, an emulsifying agent or a coating agent.
  • wetting agents such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, release agents, coating agents, sweetening, flavoring, perfuming agents, preservatives, and antioxidants may be present in the formulated agents.
  • compositions may be suitable for oral, nasal, topical (including buccal and sublingual), rectal, vaginal, aerosol and/or parenteral administration.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy.
  • the amount of composition that may be combined with a carrier material to produce a single dose may vary depending upon the subject being treated, and the particular mode of administration.
  • Formulations suitable for oral administration may be in the form of capsules, cachets, pills, tablets, lozenges (using a flavored basis, usually sucrose and acacia or tragacanth), powders, granules, or as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oil-in-water or water-in-oil liquid emulsion, or as an elixir or syrup, or as pastilles (using an inert base, such as gelatin and glycerin, or sucrose and acacia), each containing a predetermined amount of a subject composition thereof as an active ingredient.
  • Compositions of the present invention may also be administered as a bolus, electuary, or paste.
  • compositions of the disclosure may be mixed with one or more pharmaceutically acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (5) solution retarding agents, such as paraffin; (6) absorption accelerators, such as quaternary ammonium compounds; (7) wetting agents, such as
  • compositions may also comprise buffering agents.
  • Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols and the like.
  • Liquid dosage forms for oral administration may include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups, and elixirs.
  • the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, cyclodextrins and mixtures thereof.
  • inert diluents commonly used in the art, such as, for example, water or other solvents, solub
  • Suspensions in addition to the subject composition, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
  • suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
  • compositions are described as having, including, or comprising specific components, it is contemplated that compositions also consist essentially of, or consist of, the recited components.
  • processes are described as having, including, or comprising specific process steps, the processes also consist essentially of, or consist of, the recited processing steps.
  • the compounds disclosed herein can be prepared in a number of ways well known to one skilled in the art of organic synthesis.
  • the present disclosure also provides methods for treating, preventing, or ameliorating lactose intolerance or lactase deficiency by administering a pharmaceutical composition comprising one or more isolated fatty acids, e.g., a conjugated linoleic acid (CLA), for example, a trans-10, cis-12 conjugated linoleic acid isomer, a cis-9, trans-11 conjugated linoleic acid isomer, or a mixture thereof.
  • CLA conjugated linoleic acid
  • the disclosure provides pharmaceutical compositions for use in treating lactose intolerance or lactase deficiency.
  • Pharmaceutical compositions may be comprised of a disclosed isolated fatty acid, for example, a CLA, and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition may be a mixture containing a specified amount of a therapeutic compound, e.g., a therapeutically effective amount, of a therapeutic compound, for example, a therapeutically effective amount of a fatty acid (e.g., a CLA), in a pharmaceutically acceptable carrier for administering to a patient, e.g., a human, in order to treat, manage, ameliorate, and/or prevent lactose intolerance or lactase deficiency.
  • a pharmaceutically acceptable carrier for administering to a patient, e.g., a human, in order to treat, manage, ameliorate, and/or prevent lactose intolerance or lactase deficiency.
  • pharmaceutical compositions comprising a disclosed isolated fatty acid and a pharmaceutically acceptable carrier.
  • the disclosure is directed to use of a isolated fatty acid in the manufacture of a medicament for treating, managing, ameliorating, and/or preventing lactose intolerance or a lactase deficiency.
  • “Medicament,” as used herein, has essentially the same meaning as the term “pharmaceutical composition.”
  • Pharmaceutically acceptable carriers may include buffers, carriers, and excipients suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • the carrier(s) should be “acceptable” in the sense of being compatible with the other ingredients of the formulations and not deleterious to the recipient.
  • Pharmaceutically acceptable carriers include buffers, solvents, dispersion media, coatings, isotonic and absorption delaying agents, and the like, that are compatible with pharmaceutical administration. The use of such media and agents for pharmaceutically active substances is known in the art.
  • the pharmaceutical composition is administered orally and includes an enteric coating or a lipophilic coating suitable for regulating the site of absorption of the encapsulated substances within the digestive system or gut.
  • an enteric coating can include an ethylacrylate-methacrylic acid copolymer, an amino alkyl methacrylate copolymer, a methacrylic acid copolymer, a methacrylic ester copolymer, an ammonioalkyl methacrylate copolymer, a polymethacrylate, a poly(methacrylic acid-co-methyl methacrylate), hydroxypropyl-methylcellulose phthalate.
  • formulations provided herein include enteric coatings, for example, lipophilic coatings, that allow delivery of a therapeutic, for example, an isolated fatty acid, to one or more specific regions of the gastrointestinal tract.
  • formulations may include enteric coatings and reagents that allow delivery of therapeutic to the stomach, the duodenum, the jejunum, the small intestine, the large intestine, the transverse, ascending, or descending colon, the ileum, the cecum, and/or the rectum.
  • Formulations may include enteric coatings and reagents that allow release of therapeutic from a formulation for oral administration in the form of, for example, a tablet, a lozenge, or a capsule, at an approximate pH value or within a pH value range.
  • formulations provided herein may include enteric coatings and reagents that release therapeutic, for example, an isolated fatty acid, from a formulation for oral administration at a pH value of about 3, about 4, about 4.5, about 5, about 5.5, about 6, about 6.5, about 7, about 7.5, or about 8.
  • formulations provided herein may include enteric coatings and reagents that release therapeutic from a formulation for oral administration at a pH value of greater than about 3, greater than about 4, greater than about 4.5, greater than about 5, greater than about 5.5, greater than about 6, greater than about 6.5, greater than about 7, greater than about 7.5, or greater than about 8.
  • formulations of the disclosure release therapeutic from a formulation for oral administration in a pH value range of about pH 3 to about pH, about pH 4 to about pH 5, about pH 5 to about pH 6, about pH 6 to about pH 7, about pH 7 to about pH 8, about pH 8 to about pH 9, about pH 4.5 to about pH 7.5, about pH 4 to about pH 7, about pH 5 to about pH 7, about pH 5.5 to about pH 6.5, or about pH 4.5 to about pH 5.5.
  • a disclosed fatty acid and any pharmaceutical composition thereof may be administered by one or several routes, including topically, parenterally, orally, pulmonarily, intratracheally, intranasally, transdermally, or intraduodenally.
  • Parenteral administration includes subcutaneous injections, intrapancreatic administration, intravenous, intramuscular, intraperitoneal, intrasternal injection or infusion techniques.
  • a fatty acid may be administered subcutaneously to a subject.
  • a fatty acid may be administered orally to a subject.
  • a fatty acid may be administered directly to the gastrointestinal system, or specific regions of the gastrointestinal system (e.g., the ileum, colon, or rectum) via parenteral administration.
  • compositions containing a fatty acid can be presented in a dosage unit form and can be prepared by any suitable method.
  • a pharmaceutical composition should be formulated to be compatible with its intended route of administration.
  • Useful formulations can be prepared by methods well known in the pharmaceutical art. For example, see Remington's Pharmaceutical Sciences, 18th ed. (Mack Publishing Company, 1990).
  • compositions for example, are sterile. Sterilization can be accomplished, for example, by filtration through sterile filtration membranes. Where the composition is lyophilized, filter sterilization can be conducted prior to or following lyophilization and reconstitution.
  • compositions of the disclosure can be formulated for parenteral administration, e.g., formulated for injection via the intravenous, intramuscular, subcutaneous, intralesional, or intraperitoneal routes.
  • parenteral administration e.g., formulated for injection via the intravenous, intramuscular, subcutaneous, intralesional, or intraperitoneal routes.
  • the preparation of an aqueous composition such as an aqueous pharmaceutical composition containing a fatty acid, will be known to those of skill in the art in light of the present disclosure.
  • such compositions can be prepared as injectables, either as liquid solutions or suspensions; solid forms suitable for using to prepare solutions or suspensions upon the addition of a liquid prior to injection can also be prepared; and the preparations can also be emulsified.
  • the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions; formulations including sesame oil, peanut oil or aqueous propylene glycol; and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi.
  • Solutions of active compounds as free base or pharmacologically acceptable salts can be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. In addition, sterile, fixed oils may be employed as a solvent or suspending medium. For this purpose any bland fixed oil can be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid can be used (beyond their use as therapeutic agents) in the preparation of injectables.
  • Sterile injectable preparations may also be sterile injectable solutions, suspensions, or emulsions in a nontoxic parenterally acceptable diluent or solvent, for example, as solutions in 1,3-butanediol.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S.P., and isotonic sodium chloride solution.
  • a fatty acid may be suspended in a carrier fluid comprising 1% (w/v) sodium carboxymethylcellulose and 0.1% (v/v) TWEENTM 80. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • Injectable preparations for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents.
  • dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • Sterile injectable solutions of the disclosure may be prepared by incorporating a fatty acid in the required amount of the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization.
  • the preferred methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • the injectable formulations can be sterilized, for example, by filtration through a bacteria-retaining filter.
  • DMSO dimethyl methacrylate
  • Suitable preservatives for use in such a solution include benzalkonium chloride, benzethonium chloride, chlorobutanol, thimerosal and the like.
  • Suitable buffers include boric acid, sodium and potassium bicarbonate, sodium and potassium borates, sodium and potassium 10 carbonate, sodium acetate, sodium biphosphate and the like, in amounts sufficient to maintain the pH at between about pH 6 and pH 8, and for example, between about pH 7 and pH 7.5.
  • Suitable tonicity agents are dextran 40, dextran 70, dextrose, glycerin, potassium chloride, propylene glycol, sodium chloride, and the like, such that the sodium chloride equivalent of the solution is in the range 0.9 plus or minus 0.2%.
  • Suitable antioxidants and stabilizers include sodium bisulfite, sodium metabisulfite, sodium thiosulfite, thiourea and the like.
  • Suitable wetting and clarifying agents include polysorbate 80, polysorbate 20, poloxamer 282 and tyloxapol.
  • Suitable viscosity-increasing agents include dextran 40, dextran 70, gelatin, glycerin, hydroxyethylcellulose, hydroxymethylpropylcellulose, lanolin, methylcellulose , petrolatum, polyethylene glycol, polyvinyl alcohol, polyvinylpyrrolidone, carboxymethylcellulose and the like.
  • compositions suitable for oral delivery of a fatty acid e.g., tablets that include an enteric coating, e.g., a gastro-resistant coating, such that the compositions may deliver fatty acid to, e.g., the gastrointestinal tract of a patient.
  • an enteric coating e.g., a gastro-resistant coating
  • Such administration may result in a topical effect, substantially topically applying the fatty acid directly to an affected portion of the gastrointestinal tract of a patient.
  • Such administration may, in some embodiments, substantially avoid unwanted systemic absorption of a fatty acid.
  • a tablet for oral administration comprises granules (e.g., is at least partially formed from granules) that include a fatty acid, e.g., an isolated naturally occurring fatty acid, e.g., a trans-10, cis-12 conjugated linoleic acid isomer, a cis-9, trans-11 conjugated linoleic acid isomer, or a mixture of one or more conjugated linoleic acids, and one or more pharmaceutically acceptable excipients.
  • a fatty acid e.g., an isolated naturally occurring fatty acid, e.g., a trans-10, cis-12 conjugated linoleic acid isomer, a cis-9, trans-11 conjugated linoleic acid isomer, or a mixture of one or more conjugated linoleic acids, and one or more pharmaceutically acceptable excipients.
  • Such a tablet may be coated with an enteric coating.
  • Tablets provided herein may include pharmaceutically acceptable excipients such as fillers, binders, disintegrants, and/or lubricants, as well as coloring agents, release agents, coating agents, sweetening, flavoring such as wintergreen, orange, xylitol, sorbitol, fructose, and maltodextrin, and perfuming agents, preservatives and/or antioxidants.
  • pharmaceutically acceptable excipients such as fillers, binders, disintegrants, and/or lubricants, as well as coloring agents, release agents, coating agents, sweetening, flavoring such as wintergreen, orange, xylitol, sorbitol, fructose, and maltodextrin, and perfuming agents, preservatives and/or antioxidants.
  • provided pharmaceutical formulations include an intra-granular phase that includes a fatty acid, e.g., an isolated naturally occurring fatty acid, e.g., a trans-10, cis-12 conjugated linoleic acid isomer, a cis-9, trans-11 conjugated linoleic acid isomer, or a mixture of one or more conjugated linoleic acids, and a pharmaceutically acceptable salt, e.g., a disclosed fatty acid, e.g., an isolated naturally occurring fatty acid, e.g., a trans-10, cis-12 conjugated linoleic acid isomer, a cis-9, trans-11 conjugated linoleic acid isomer, or a mixture of one or more conjugated linoleic acids, and a pharmaceutically acceptable filler.
  • a fatty acid e.g., an isolated naturally occurring fatty acid, e.g., a trans-10, cis-12 conjugated lin
  • a disclosed fatty acid and a filler may be blended together, optionally, with other excipients, and formed into granules.
  • the intragranular phase may be formed using wet granulation, e.g. a liquid (e.g., water) is added to the blended fatty acid compound and filler, and then the combination is dried, milled and/or sieved to produce granules.
  • wet granulation e.g. a liquid (e.g., water) is added to the blended fatty acid compound and filler, and then the combination is dried, milled and/or sieved to produce granules.
  • a liquid e.g., water
  • provided formulations include an extra-granular phase, which may include one or more pharmaceutically acceptable excipients, and which may be blended with the intragranular phase to form a disclosed formulation.
  • a disclosed formulation may include an intragranular phase that includes a filler.
  • exemplary fillers include, but are not limited to, cellulose, gelatin, calcium phosphate, lactose, sucrose, glucose, mannitol, sorbitol, microcrystalline cellulose, pectin, polyacrylates, dextrose, cellulose acetate, hydroxypropylmethyl cellulose, partially pre-gelatinized starch, calcium carbonate, and others including combinations thereof.
  • a disclosed formulation may include an intragranular phase and/or a extragranular phase that includes a binder, which may generally function to hold the ingredients of the pharmaceutical formulation together.
  • binders of the disclosure may include, but are not limited to, the following: starches, sugars, cellulose or modified cellulose such as hydroxypropyl cellulose, lactose, pre-gelatinized maize starch, polyvinyl pyrrolidone, hydroxypropyl cellulose, hydroxypropylmethyl cellulose, low substituted hydroxypropyl cellulose, sodium carboxymethyl cellulose, methyl cellulose, ethyl cellulose, sugar alcohols and others including combinations thereof.
  • Formulations of the disclosure may include a disintegrant such as but are not limited to, starch, cellulose, crosslinked polyvinyl pyrrolidone, sodium starch glycolate, sodium carboxymethyl cellulose, alginates, corn starch, crosmellose sodium, crosslinked carboxymethyl cellulose, low substituted hydroxypropyl cellulose, acacia, and others including combinations thereof.
  • a disintegrant such as but are not limited to, starch, cellulose, crosslinked polyvinyl pyrrolidone, sodium starch glycolate, sodium carboxymethyl cellulose, alginates, corn starch, crosmellose sodium, crosslinked carboxymethyl cellulose, low substituted hydroxypropyl cellulose, acacia, and others including combinations thereof.
  • a disintegrant such as but are not limited to, starch, cellulose, crosslinked polyvinyl pyrrolidone, sodium starch glycolate, sodium carboxymethyl cellulose, alginates, corn starch, crosmellose sodium, crosslinked carb
  • a provided formulation includes an intra-granular phase comprising a fatty acid and excipients chosen from: mannitol, microcrystalline cellulose, hydroxypropylmethyl cellulose, and sodium starch glycolate or combinations thereof, and an extra-granular phase comprising one or more of: microcrystalline cellulose, sodium starch glycolate, and magnesium stearate or mixtures thereof.
  • a provided formulation may include a lubricant, e.g., an extra-granular phase may contain a lubricant.
  • Lubricants include but are not limited to talc, silica, fats, stearin, magnesium stearate, calcium phosphate, silicone dioxide, calcium silicate, calcium phosphate, colloidal silicon dioxide, metallic stearates, hydrogenated vegetable oil, corn starch, sodium benzoate, polyethylene glycols, sodium acetate, calcium stearate, sodium lauryl sulfate, sodium chloride, magnesium lauryl sulfate, talc, and stearic acid.
  • a pharmaceutical formulation comprises an enteric coating, for example, a lipophilic coating.
  • enteric coatings create a barrier for the oral medication that controls the location at which the drug is absorbed along the digestive tract.
  • Enteric coatings may include a polymer that disintegrates at different rates according to pH.
  • Enteric coatings may include for example, cellulose acetate phthalate, methyl acrylate-methacrylic acid copolymers, cellulose acetate succinate, hydroxylpropylmethyl cellulose phthalate, methyl methacrylate-methacrylic acid copolymers, ethylacrylate-methacrylic acid copolymers, methacrylic acid copolymer type C, polyvinyl acetate-phthalate, and cellulose acetate phthalate.
  • enteric coatings include Opadry® AMB, Acryl-EZE®, Eudragit® grades.
  • an enteric coating may comprise about 5% to about 10%, about 5% to about 20%, 8 to about 15%, about 8% to about 20%, about 10% to about 20%, or about 12 to about 20%, or about 18% of a tablet by weight.
  • enteric coatings may include an ethylacrylate-methacrylic acid copolymer.
  • a tablet that comprises or consists essentially of about 0.5% to about 70%, e.g. about 0.5% to about 10%, or about 1% to about 20%, by weight of a fatty acid or a pharmaceutically acceptable salt thereof.
  • a tablet can include, for example, about 0.5% to about 60% by weight of mannitol, e.g., about 30% to about 50% by weight mannitol, e.g., about 40% by weight mannitol; and/or about 20% to about 40% by weight of microcrystalline cellulose, or about 10% to about 30% by weight of microcrystalline cellulose.
  • a disclosed tablet may comprise an intragranular phase that includes about 30% to about 60%, e.g.
  • a pharmaceutically acceptable tablet for oral use may include about 0.5% to 10% by weight of a disclosed fatty acid, e.g., a CLA or a pharmaceutically acceptable salt thereof, about 30% to 50% by weight mannitol, about 10% to 30% by weight microcrystalline cellulose, and an enteric coating comprising an ethylacrylate-methacrylic acid copolymer.
  • a disclosed fatty acid e.g., a CLA or a pharmaceutically acceptable salt thereof
  • enteric coating comprising an ethylacrylate-methacrylic acid copolymer.
  • a pharmaceutically acceptable tablet for oral use may comprise an intra-granular phase, comprising about 5 to about 10% by weight of a fatty acid, e.g., a CLA, or a pharmaceutically acceptable salt thereof, about 40% by weight mannitol, about 8% by weight microcrystalline cellulose, about 5% by weight hydroxypropylmethyl cellulose, and about 2% by weight sodium starch glycolate; an extra-granular phase comprising about 17% by weight microcrystalline cellulose, about 2% by weight sodium starch glycolate, about 0.4% by weight magnesium stearate; and an enteric coating over the tablet comprising an ethylacrylate-methacrylic acid copolymer.
  • a fatty acid e.g., a CLA, or a pharmaceutically acceptable salt thereof
  • the pharmaceutical composition may contain an enteric coating comprising about 13% or about 15%, 16%, 17% or 18% by weight, e.g., AcyrlEZE® (see, e.g., PCT Publication No. WO2010/054826, which is hereby incorporated by reference in its entirety).
  • an enteric coating comprising about 13% or about 15%, 16%, 17% or 18% by weight, e.g., AcyrlEZE® (see, e.g., PCT Publication No. WO2010/054826, which is hereby incorporated by reference in its entirety).
  • a tablet may have a dissolution profile, e.g. when tested in a USP/EP Type 2 apparatus (paddle) at 100 rpm and 37° C. in a phosphate buffer with a pH of 7.2, of about 50% to about 100% of the fatty acid releasing after about 120 minutes to about 240 minutes, for example after 180 minutes.
  • a tablet may have a dissolution profile, e.g. when tested in a USP/EP Type 2 apparatus (paddle) at 100 rpm and 37° C. in diluted HC1 with a pH of 1.0, where substantially none of the fatty acid is released after 120 minutes.
  • a tablet provided herein may have a dissolution profile, e.g. when tested in USP/EP Type 2 apparatus (paddle) at 100 rpm and 37° C. in a phosphate buffer with a pH of 6.6, of about 10% to about 30%, or not more than about 50%, of the fatty acid releasing after 30 minutes.
  • a dissolution profile e.g. when tested in USP/EP Type 2 apparatus (paddle) at 100 rpm and 37° C. in a phosphate buffer with a pH of 6.6, of about 10% to about 30%, or not more than about 50%, of the fatty acid releasing after 30 minutes.
  • Formulations when orally administered to the patient may result in minimal plasma concentration of the fatty acid in the patient.
  • disclosed formulations when orally administered to a patient, topically deliver to the colon or rectum of a patient, e.g., to an affected or diseased site of a patient.
  • methods provided herein may further include administering at least one other agent that is directed to treatment of diseases and disorders disclosed herein.
  • contemplated other agents may be co-administered (e.g., sequentially or simultaneously).
  • immunosuppressive agents including glucocorticoids, cytostatics, antibodies, agents acting on immunophilins, interferons, opioids, TNF binding proteins, mycophenolate, and small biological agents.
  • contemplated immunosuppressive agents include, but are not limited to: tacrolimus, cyclosporine, pimecrolimus, sirolimus, everolimus, mycophenolic acid, fingolimod, dexamethasone, fludarabine, cyclophosphamide, methotrexate, azathioprine, leflunomide, teriflunomide, anakinra, anti-thymocyte globulin, anti-lymphocyte globulin, muromonab-CD3, afutuzumab, rituximab, teplizumab, efalizumab, daclizumab, basiliximab, adalimumab, inflixima
  • Exemplary formulations include dosage forms that include or consist essentially of about 35 mg to about 500 mg of a fatty acid.
  • formulations that include about 35 mg, 40 mg, 50 mg, 60 mg, 70 mg, 80 mg, 90 mg, 100 mg, 110 mg, 120 mg, 130 mg, 140 mg, 150 mg, 160 mg, 170 mg, 180 mg, 190 mg, 200 mg, or 250 mg of a fatty acid are provided herein.
  • a formulation may include about 40 mg, 80 mg, or 160 mg of a fatty acid.
  • a formulation may include at least 100 ⁇ g of a fatty acid.
  • formulations may include about 0.1 mg, 0.2 mg, 0.3 mg, 0.4 mg, 0.5 mg, 1 mg, 5 mg, 10 mg, 15 mg, 20 mg, or 25 mg of a fatty acid.
  • the amount administered will depend on variables such as the type and extent of disease or indication to be treated, the overall health and size of the patient, the in vivo potency of the fatty acid, the pharmaceutical formulation, and the route of administration.
  • the initial dosage can be increased beyond the upper level in order to rapidly achieve the desired blood-level or tissue level. Alternatively, the initial dosage can be smaller than the optimum, and the dosage may be progressively increased during the course of treatment.
  • Human dosage can be optimized, e.g., in a conventional Phase I dose escalation study designed to run from 40 mg to 160 mg.
  • Dosing frequency can vary, depending on factors such as route of administration, dosage amount and the disease being treated. Dosing frequencies can include once per day, twice per day, 3 times per day, 4 times per day, 5 times per day, 6 times per day, 7 times per day, 8 times per day, 9 times per day, 10 times per day, more than 10 times per day, once per week, once every two weeks, once per month, and as needed. In some embodiments, dosing is once per day for 7 days.
  • Caco-2 (colonic adenocarcinoma) cells were grown in Dulbecco's Modified Eagle's Medium (DMEM, Invitrogen, Life Technologies, Cergy-Pontoise, France) supplemented with 20% foetal calf serum (FCS, Dutscher, Brumath, France), 1% penicillin-streptomycin (5 ml/l) (Invitrogen, Life technologies) and 1% non-essential amino acids (5 ml/l) (Invitrogen, Life technologies). All cell lines were cultured as confluent monolayers at 37° C. in a controlled, 5% CO 2 atmosphere.
  • DMEM Dulbecco's Modified Eagle's Medium
  • FCS foetal calf serum
  • FCS foetal calf serum
  • penicillin-streptomycin 5 ml/l
  • non-essential amino acids 5 ml/l
  • RNA samples were lysed with lysis buffer (RA1, Macherey-Nagel) containing 1% ⁇ -mercaptoethanol.
  • Total RNA was extracted with a Nucleospin RNA kit (Macherey-Nagel, Hoerdt, France). After RNAse inactivation, total RNA was cleaned of genomic DNA traces by DNAse treatment and eluted in RNAse-free, DEPC-water. The purity of the RNA was evaluated by UV spectroscopy on a Nanodrop system (Nyxor Biotech, Paris, France) from 220 to 350 nm. Before microarray experiments, RNAs were also profiled on an Agilent 2100 bioanalyzer. One ⁇ g of total RNA with a minimum concentration of 50 ng/ ⁇ l was used in the microarray and qRT-PCR analysis.
  • Dual-colour gene expression microarrays were used to compare the cRNA from the samples. 44,000 genes were screened.
  • the RNAs from the samples were first reverse-transcribed into cDNA (Affinity-Script RT, Agilent), which were then used as the substrate for the synthesis and amplification of cRNA by T7 RNA polymerase in the presence of cyanine 3-CTP for the CTL sample (green fluorescence) and cyanine 5-CTP for the PPAR- ⁇ y modulator sample (red fluorescence).
  • the two-labelled cRNAs were mixed, hybridized on the same array (G4851A Agilent 8 ⁇ 44K) and then scanned (with an Agilent G2505B scanner).
  • qPCR quantitative PCR
  • RNA was reverse-transcribed into cDNA using the High Capacity cDNA Archive kit (Applied Biosystems). Amplification was performed using an ABI PRISM 7000 sequence detection system (Applied Biosystem) using Power SYBR® Green PCR master Mix (Applied Biosystem). Primer pairs for each human transcript were chosen with qPrimer depot software (at primerdepot.nci.nih.gov.). Quantification of qPCR signals was performed using ⁇ Ct relative quantification method using GAPDH as a reference gene for human and rat samples and ⁇ -Actin for mouse samples. Values were represented in terms of relative quantity of mRNA level variation or fold increase compared to control conditions.
  • LCT protein expression level was determined by immunoprecipitation followed by Western Blotting analysis. Briefly, total proteins were extracted from Caco-2 cells using a RIPA Buffer containing 25 mM Tris-HCl pH 7.6, 150 mM NaCl, 1% NP-40, 1% Sodium deoxycholate, 0.1% Sodium Dodecyl sulphate supplemented with classical protease-inhibitor cocktail. 250 ⁇ g of total protein were immunoprecipitated with 2 ⁇ g of a specific antibody against Lactase (Santa Cruz) overnight at 4° C. The immunoprecipitated proteins were coated with protein A/G Agarose beads (Santa Cruz) and mixed gently for 4 hours at 4° C.
  • Membranes were then incubated with secondary horseradish peroxidase-conjugated antibodies (anti-rabbit (Jackson ImmunoResearch) and anti-mouse (Sigma), 1:10000 for 1 hour at room temperature) and finally revealed with chemiluminescent substrate according to the manufacturer's protocol (ECL; Millipore Corporation). Membranes were exposed to autoradiography films (Hyperfilm; Amersham Biosciences).
  • Lactase activity was evaluated by using a glucose oxidase method (Glucose Assay Kit, Sigma). This lactase assay is based on the measurement of the amount of glucose produced following the action of lactase by incubating samples with a lactose buffer solution (0.056 mol/l lactose in a 0.1 mol/l Na-maleate buffer).
  • lactose buffer solution 0.056 mol/l lactose in a 0.1 mol/l Na-maleate buffer.
  • lactase activity was determined directly from the cell monolayer. After extensive washing, the cell monolayer was incubated with lactose buffer for one hour at 37° C. The supernatant was recovered, 50 ⁇ l were diluted with 100 ⁇ l of glucose oxidase reagent and incubated at 37° C. for 1 hour.
  • the reaction was stopped with 100 ⁇ l of H 2 SO 4 and read by spectrophotometry at 450 nm.
  • tissue samples were first dounce-homogenized in 0.9% NaCl on crushed ice. These homogenates were then diluted in 0.9% NaCl ( 1/500) and 50 ⁇ l of dilution were incubated with lactose buffer and used to determine lactase activity.
  • the background attributed to the remaining glucose in the samples was measured by incubating cells or cell extracts in lactose-free buffer.
  • Glucose uptake was evaluated by using the glucose uptake colorimetric assay kit (Sigma-Aldrich) according to the manufacturer's instructions. Briefly, Caco-2 cells were seeded into a 96-well plate at a density of 30,000 cells per well. Cells were serum deprived for 16 hours prior to stimulation in order to synchronize the cells. Cells were treated with GED (1 mM) or pioglitazone (1 ⁇ M) for 24 h.
  • KRPH buffer Krebs-Ringer-Phosphate-HEPES (KRPH) Buffer—20 mM HEPES, 5 mM KH2PO4, 1 mM MgSO4, 1 mM CaCl2, 136 mM NaCl, and 4.7 mM KCl, pH 7.4) containing 2% BSA for 40 minutes. 10 ⁇ l of 2-deoxyglucose (2-DG; 10 mM) was then added and incubation was continued for 20 minutes.
  • 2-DG 2-deoxyglucose
  • 2-DG is taken up by the cells and phosphorylated by hexokinase to 2-DG6P, which cannot be further metabolized and accumulates in cells. Following incubation, cells were washed 3 times with PBS and lysed with 80 ⁇ l of the extraction buffer provided. The amount of 2-DG6P (which is directly proportional to glucose uptake by the cells) was determined by a colorimetric detection assay according to the manufacturer's protocol.
  • PPAR ⁇ knockdown IECs were obtained using the pSUPER.retro system (OligoEngine). Forward and reverse target sequences corresponding to nucleotides 105-123 of the human PPAR ⁇ mRNA (5′-GCCCTTCACTACTGTTGAC-3′ (SEQ ID NO: 1)) were cloned into the BgllI/XhoI restriction sites of the pSUPERretro vector (pRS) giving the ShPPAR construct.
  • pRS pSUPERretro vector
  • a negative control pRS plasmid containing the sequence 5′-ACGCTGAGTACTTCGAAAT-3′ (SEQ ID NO: 2) targeted against the luciferase gene was also generated (ShLuc construct).
  • Both constructions were transfected in Caco-2 cells using Nucleofector technology from Amaxa Biosystems, according to the manufacturer's protocol. Stably transfected clones were selected 24 h post-transfection with complete culture medium supplemented with puromycin (5 ⁇ g/ml). The silencing of PPAR ⁇ expression was checked by quantitative RT-PCR and western-blot analysis. Once established, ShPPAR and ShLuc cell lines were maintained in complete medium supplemented with 2.5 ⁇ g/ml puromycin.
  • a 321 bp genomic fragment (corresponding to the first 321 bp upstream to the transcription start site of the human lactase gene) was cloned in the pGL4-Luc reporter vector using XhoI and Hind III restriction sites introduced in “Hs-Prom-0.3 Kb sens” and “Hs-Prom-0.3 Kb anti-sens” oligonucleotides respectively.
  • This construct and the empty vector control were transiently transfected in Caco-2 cells using NucleofectorTM Technology. Six hours post-transfection, cells were treated with PPAR ⁇ modulator for 12 hours. Luciferase activity was measured using the luciferase assay kit (Promega) in a Wallac Victor2 TM 1420 multilabel counter (Perkin Elmer).
  • PPAR ⁇ onto the LCT gene promoter was studied by Chromatin immunoprecipitation (ChIP) experiments in Caco-2 cells (5 ⁇ 10 6 cells) stimulated for 24 hours with 1 mM GED in 100 mm cell culture petri dishes.
  • Caco-2 cells were synchronized by the addition of serum-free medium for 16 hours and then stimulated for 24 hours using the protocol described previously. Cells were then rinsed with PBS and the protein-DNA complexes were fixed by adding 1% PFA for 30 minutes at room temperature. This binding was stopped by the addition of glycine (0.125M). Cells were collected by scraping in the presence of cold PBS and protease inhibitors (Sigma).
  • the cell pellet obtained by centrifugation was taken up in 300 ⁇ l SDS buffer (1% SDS, 10 mM EDTA, 50 mM Tris-HCl pH 8, protease inhibitors) and sonicated (Diagenode, BioruptorUCD200TM-EX) for 30 seconds, followed by 30 seconds resting time.
  • 125 ⁇ L of crosslinked sonicated sample was diluted with 225 ⁇ l of IP buffer (1% triton X-100, 150 mM NaCl, 2 mM EDTA, 20 mM Tris-HCl pH 8.1 and protease inhibitors) and pre-cleared for four hours by adding 40 ⁇ L of protein A/G beads (50% slurry protein A/G Sepharose, Clinisciences) and 5 ⁇ g of salmon sperm DNA (Invitrogen).
  • protein A/G beads 50% slurry protein A/G Sepharose, Clinisciences
  • 5 ⁇ g of salmon sperm DNA Invitrogen.
  • Complexes were immunoprecipitated with specific anti-PPAR ⁇ antibodies (C26H12 rabbit monoclonal antibody, CellSignaling) by incubation overnight at 4° C. under rotation.
  • Immune complexes were recovered by adding 40 ⁇ L of protein A/G Sepharose (50%) plus 2 ⁇ g salmon sperm DNA and incubated for four hours at 4° C.
  • the beads were washed twice in wash buffer 1 (0.1% SDS, 1% Triton X-100, 150 mM NaCl, 0.1% Deoxycholate, 1 mM EGTA, 2 mM EDTA, 20 mM Tris HCl pH 8.0), twice in wash buffer 2 (0.1% SDS, 1% Triton X-100, 500 mM NaCl, 0.1% Deoxycholate, 1 mM EGTA, 2 mM EDTA, 20 mM Tris HCl pH 8.0), once in wash buffer 3 (0.25 mM LiCl, 0.5% Deoxycholate, 0.5% NP40, 0.5 mM EGTA, 1 mM EDTA, 10 mM Tris HCl pH 8.0) and 3 times in wash buffer 4 (1 mM
  • the co-immunoprecipitated DNA was then extracted with 150 ⁇ l of extraction buffer (0.1M NaHCO3, 1% SDS). Cross-linking was reversed overnight at 65° C. DNA was then purified using the PCR Clean-up kit (Macherey-Nagel) and analyzed by PCR.
  • mice and rats were housed 5 animals/cage and 3 animals/cage, respectively, in a specific pathogen-free facility, in an air-conditioned room with controlled temperature (22 ⁇ 1° C.), humidity (65-70%), and 12 h light/12 h dark cycles. Animals were fed with standard laboratory chow (except when indicated) and were provided with autoclaved tap water ad libitum. Animals were acclimatized for at least 1 week before entering the study.
  • Proximal intestine samples from knockout mice harbouring a specific PPAR- ⁇ deletion in IEC were provided by Prof. Daniel Metzger (Institute of Genetics and Molecular and Cellular Biology IGBMC (Inserm/CNRS/University of France)).
  • SCFA were extracted and measured as described in Momose, Y., et al. “Studies on antidiabetic agents X. Synthesis and biological activities of pioglitazone and related compounds.” Chem Pharm Bull (Tokyo) 39, 1440-1445 (1991), which is incorporated herein by reference.
  • LCT gene was the leading gene upregulated following stimulation with 1 mM GED, 30 mM GED, and 1 ⁇ M Pio (Table 1). LCT gene expression was significantly increased in response to 1 mM GED (5.28-fold ⁇ 0.55; P ⁇ 0.05), 30 mM GED (8.28-fold ⁇ 1.7; P ⁇ 0.05), and by Pio (17.93-fold ⁇ 5.1; P ⁇ 0.05) compared to unstimulated cells.
  • LCT activity was also evaluated following stimulation of Caco-2 cells with 1 mM GED, 30 mM GED, or 30 mM 5-ASA.
  • a significant increase in LCT activity was observed relative to control cells (CTRL) following stimulation with 1 mM or 30 mM GED, but not with 30 mM 5-ASA ( FIG. 4C ; CTRL v. 1 mM GED, p ⁇ 0.005; CTRL v. 30 mM GED, p ⁇ 0.005).
  • CC13910 and GG22018 are linked to hypolactasia.
  • the homozygous CC13910 and GG22018 genotypes are associated with the lactase non-persistent phenotype.
  • the genotype of Caco-2 cells is CC13910 and GG22018, suggesting that PPAR ⁇ modulators may be able to control LCT gene expression in lactase non-persistent individuals.
  • PPAR ⁇ is able to bind DNA as a heterodimer with another nuclear receptor RXR.
  • the heterodimer PPAR ⁇ -RXR recognizes short dimeric palindromic sequences (consensus sequence AGGTCA or TGACCT) spaced by one nucleotide (known as direct repeat 1 (DR1)) or two nucleotides (known as direct repeat 2 (DR2)), which define the PPRE.
  • DR1 direct repeat 1
  • DR2 direct repeat 2
  • ChIP Chromatin immunoprecipitation
  • a genomic fragment containing this PPAR ⁇ -bound DR2 sequence was cloned upstream of the luciferase gene sequence into a pGL4 vector (pGL4Luc Prom LCT construct) and tested in a reporter gene assay in Caco-2 cells.
  • luciferase activity significantly increased in the presence of GED stimulation (GED) compared to untreated cells (CTL; FIG. 11 ), indicating that GED stimulated PPAR ⁇ binding to the DR2 sequence and triggered increased luciferase expression.
  • LCT mRNA expression was significantly decreased in the proximal part of the small intestine of PPAR ⁇ ⁇ IEC KO mice compared to CTRL animals ( FIG. 14 ). This result demonstrates that LCT expression in the proximal small intestine was dependent upon PPAR ⁇ expression.
  • LCT mRNA expression and PPAR ⁇ mRNA expression were significantly increased in the duodenum and jejunum of unweaned wild-type Sprague-Dawley rats compared to their weaned counterparts ( FIG. 15A ). Increased PPAR ⁇ and LCT mRNA expression were also significantly correlated in the duodenum and jejunum of unweaned rats ( FIGS. 15B and 15C ). These results demonstrate that increased PPAR ⁇ and LCT expression were significantly correlated in the proximal gut of unweaned rats.
  • Rats were sacrificed at day 4, and rats treated with GED presented a significant decrease of caecum volume compared to untreated rats (data not shown), together with a significant decrease in concentration of short-chain fatty acids (SCFA), which corresponds to the ceacal end product of lactose fermentation ( FIG. 18C ).
  • SCFA short-chain fatty acids
  • Example 5 CLA Induces Lactase Gene Expression and Activity in an Intestinal Epithelial Cell Line
  • CLA also significantly increased LCT activity in Caco-2 cells at concentrations of 100 mM or more, and 1 mM CLA induced a 2-fold greater increase in LCT activity over control levels, compared to 1 mM GED ( FIG. 19C ).
  • CLA-dependent induction of LCT expression and activity was strongly compromised in PPAR ⁇ knock-down cells ( FIGS. 20A and 20B ). These results demonstrate that a natural PPAR ⁇ agonist was able to induce LCT expression and activity in an intestinal epithelial cell line, and that the observed increases in LCT mRNA expression and LCT activity were dependent upon PPAR ⁇ expression. These results also strongly suggest that PPAR ⁇ modulators naturally present in food might be promising for the management of lactose intolerance.
  • an element or component is said to be included in and/or selected from a list of recited elements or components, it should be understood that the element or component can be any one of the recited elements or components, or the element or component can be selected from a group consisting of two or more of the recited elements or components.
  • an element means one element or more than one element.
  • molecular weight is provided and not an absolute value, for example, of a polymer, then the molecular weight should be understood to be an average molecule weight, unless otherwise stated or understood from the context.
  • C 1-6 alkyl is specifically intended to individually disclose C 1 , C 2 , C 3 , C 4 , C 5 , C 6 , C 1 - C 6 , C 1 -C 5 , C 1 -C 4 , C 1 -C 3 , C 1 -C 2 , C 2 -C 6 , C 2 -C 5 , C 2 -C 4 , C 2 -C 3 , C 3 -C 6 , C 3 -C 5 , C 3 -C 4 , C 4 -C 6 , C 4 -C 5 , and C 5 -C 6 alkyl.
  • an integer in the range of 0 to 40 is specifically intended to individually disclose 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, and 40, and an integer in the range of 1 to 20 is specifically intended to individually disclose 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, and 20.
  • phrases “optionally substituted with 1-5 substituents” is specifically intended to individually disclose a chemical group that can include 0, 1, 2, 3, 4, 5, 0-5, 0-4, 0-3, 0-2, 0-1, 1-5, 1-4, 1-3, 1-2, 2-5, 2-4, 2-3, 3-5, 3-4, and 4-5 substituents.
  • compositions specifying a percentage are by weight unless otherwise specified. Further, if a variable is not accompanied by a definition, then the previous definition of the variable controls.
US16/078,555 2016-02-26 2017-02-27 Methods of treating lactose intolerance Abandoned US20190046490A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/078,555 US20190046490A1 (en) 2016-02-26 2017-02-27 Methods of treating lactose intolerance

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201662300376P 2016-02-26 2016-02-26
US16/078,555 US20190046490A1 (en) 2016-02-26 2017-02-27 Methods of treating lactose intolerance
PCT/EP2017/054526 WO2017144725A1 (en) 2016-02-26 2017-02-27 Methods of treating lactose intolerance

Publications (1)

Publication Number Publication Date
US20190046490A1 true US20190046490A1 (en) 2019-02-14

Family

ID=58162625

Family Applications (1)

Application Number Title Priority Date Filing Date
US16/078,555 Abandoned US20190046490A1 (en) 2016-02-26 2017-02-27 Methods of treating lactose intolerance

Country Status (6)

Country Link
US (1) US20190046490A1 (de)
EP (1) EP3419616A1 (de)
JP (1) JP7125350B2 (de)
CA (1) CA3014575A1 (de)
MA (1) MA43682A (de)
WO (1) WO2017144725A1 (de)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10959970B2 (en) 2009-02-16 2021-03-30 Nogra Pharma Limited Methods of treating hair related conditions
US11046641B2 (en) 2012-02-09 2021-06-29 Nogra Pharma Limited Methods of treating fibrosis
US11905232B2 (en) 2019-02-08 2024-02-20 Nogra Pharma Limited Process of making 3-(4′-aminophenyl)-2-methoxypropionic acid, and analogs and intermediates thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120100223A1 (en) * 2008-04-21 2012-04-26 Asha Nutrition Sciences, Inc. Lipid-containing compositions and methods of use thereof

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB8302708D0 (en) * 1983-02-01 1983-03-02 Efamol Ltd Pharmaceutical and dietary composition
CA2313626A1 (en) * 1997-12-12 1999-06-17 Purdue Research Foundation Methods and compositions for treating diabetes
US7015249B1 (en) * 1997-12-12 2006-03-21 Purdue Research Foundation Methods and compositions for treating diabetes
WO2003043569A2 (en) * 2001-11-16 2003-05-30 Nutrition 21, Inc. Trans-10, cis-12 conjugated linoleic acid isomer for the treatment of diseases
WO2007096148A1 (en) * 2006-02-23 2007-08-30 Lipid Nutrition B.V. Immunoregulation
IE20070129A1 (en) * 2007-02-28 2008-12-24 Giuliani Int Ltd Ppar-gamma agonists stimulate enteric defensin expression
CA3085272A1 (en) 2008-11-13 2010-05-20 Nogra Pharma Limited Antisense compositions and methods of making and using same
EP2844242A1 (de) * 2012-04-18 2015-03-11 Nogra Pharma Limited Verfahren zur behandlung lactoseintoleranz
BR112015023849A2 (pt) * 2013-03-26 2017-07-18 Lipid Therapeutics Gmbh formulação farmacêutica que compreende fosfatidilcolina para o tratamento de colite ulcerativa

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120100223A1 (en) * 2008-04-21 2012-04-26 Asha Nutrition Sciences, Inc. Lipid-containing compositions and methods of use thereof

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10959970B2 (en) 2009-02-16 2021-03-30 Nogra Pharma Limited Methods of treating hair related conditions
US11046641B2 (en) 2012-02-09 2021-06-29 Nogra Pharma Limited Methods of treating fibrosis
US11753365B2 (en) 2012-02-09 2023-09-12 Nogra Pharma Limited Methods of treating fibrosis
US11905232B2 (en) 2019-02-08 2024-02-20 Nogra Pharma Limited Process of making 3-(4′-aminophenyl)-2-methoxypropionic acid, and analogs and intermediates thereof

Also Published As

Publication number Publication date
WO2017144725A1 (en) 2017-08-31
EP3419616A1 (de) 2019-01-02
MA43682A (fr) 2018-11-28
CA3014575A1 (en) 2017-08-31
JP2019510745A (ja) 2019-04-18
JP7125350B2 (ja) 2022-08-24

Similar Documents

Publication Publication Date Title
Zufiria et al. ALS: A bucket of genes, environment, metabolism and unknown ingredients
US7981915B2 (en) Methods for modulating PPAR biological activity for the treatment of diseases caused by mutations in the CFTR gene
Stojancevic et al. The impact of farnesoid X receptor activation on intestinal permeability in inflammatory bowel disease
US11858888B2 (en) Compounds, compositions, and methods for the treatment of inflammatory, degenerative, and neurodegenerative diseases
JP2021107404A (ja) リソソーム蓄積症治療のための組成物及び方法
Festuccia et al. PPARγ activation attenuates glucose intolerance induced by mTOR inhibition with rapamycin in rats
JP7125350B2 (ja) ラクトース不耐性を処置する方法
US9682050B2 (en) Methods of treating lactose intolerance
Edmunds et al. Liver-specific Prkn knockout mice are more susceptible to diet-induced hepatic steatosis and insulin resistance
Danesi et al. Food-derived bioactives as potential regulators of the IL-12/IL-23 pathway implicated in inflammatory bowel diseases
De Lisle et al. Lubiprostone ameliorates the cystic fibrosis mouse intestinal phenotype
Qiu et al. Preventative effects of selenium-enriched Bifidobacterium longum on irinotecan-induced small intestinal mucositis in mice
Chu et al. The selective PPAR-delta agonist seladelpar reduces ethanol-induced liver disease by restoring gut barrier function and bile acid homeostasis in mice
US20220119768A1 (en) Method for removing senescent cell, and method for preparing senescent cell
Okada et al. Intermittent fasting prompted recovery from dextran sulfate sodium-induced colitis in mice
Wu et al. Epigallocatechin-3-gallate sensitizes IFN-γ-stimulated CD4+ T cells to apoptosis via alternative activation of STAT1
WO2001068085A1 (en) A method for stimulation of defensin production
US20020006942A1 (en) Methods of treating liver disorders and disorders associated with liver function
US20230165849A2 (en) Use of pridopidine for treating rett syndrome
TWI681769B (zh) 含src同源區2蛋白酪胺酸磷酸酶-1增效劑用於改善纖維化之用途
Sieper et al. THU0275 Sustained efficacy of adalimumab in patients with non-radiographic axial spondyloarthritis: Week 68 results from ability 1
Lin et al. MiR-155 protects against sepsis-induced cardiomyocyte apoptosis via activation of NO/cGMP signaling pathway by eNOS
CN116159048A (zh) 岩藻黄素在制备治疗nlrp3诱导的肝细胞焦亡相关疾病的药物中的应用
US20220160663A1 (en) Immunosuppressant Comprising TSAHC or a Pharmaceutically Acceptable Salts Thereof as an Active Ingredient
Chen et al. Metabolic reprogramming and immunity in cancer

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: APPLICATION DISPATCHED FROM PREEXAM, NOT YET DOCKETED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION